21
March 14, 2017 Dockets Management Branch (HFA-305) Food and Drug Administration 5630 Fishers Lane, Rm. 1061 Rockville, MD 20852 Re: Docket No. FDA-2016-D-4460: Multiple Endpoints in Clinical Trials Dear Sir/Madam: The Biotechnology Innovation Organization (BIO) thanks the Food and Drug Administration (FDA) for the opportunity to submit comments on the Draft Guidance “Multiple Endpoints in Clinical Trials” (Draft Guidance). BIO is the world's largest trade association representing biotechnology companies, academic institutions, state biotechnology centers and related organizations across the United States and in more than 30 other nations. BIO members are involved in the research and development of innovative healthcare, agricultural, industrial, and environmental biotechnology products. This important Draft Guidance was well written and provides instrumental guidance and recommendations to handle major multiplicity problems in clinical trials. BIO believes that the Draft Guidance is a very helpful document to Sponsors. However, we note that the Draft Guidance fails to address a few points that are worthy of attention: multiplicity adjustments at the interim analysis (either directly in this Draft Guidance or via reference to ICH E9 or the Guidance for Industry Adaptive Design Clinical Trials for Drugs and Biologics); multiplicity adjustment on safety endpoints when assessing important safety signals as part of the objectives of the trial (e.g., with p-values reported) to control the overall false discovery rate of safety signals; elucidation of the calculation methods to be used with the truncated Holm and truncated Hochberg procedures (e.g., for the truncated Hochberg example, the α passed to the next level); and basic principles, such as closed test or partition principles, which many of the multiple comparison methods in the guidance are based upon. BIO further believes that it would be helpful if the Draft Guidance stated at the beginning, perhaps more clearly and emphatically than it does now, the areas that are mentioned in other sections as being out of scope. It would also be helpful to explain why these areas are outside of the scope. For example, the Draft Guidance states that multiplicity in interim analysis is not within the scope of the document (lines 180-184). This is something that should also be stated in the introduction. Simulation based multiplicity adjustment is another area that is not included in the guidance. If related topics are outside the scope but they have been the subject of previous FDA guidance, such guidance should be referenced.

March 14, 2017 Food and Drug Administration … · Food and Drug Administration 5630 Fishers Lane, ... BIO Comments on Multiple Endpoints in Clinical Trials FDA Docket: ... incorporation

  • Upload
    lekiet

  • View
    216

  • Download
    0

Embed Size (px)

Citation preview

March 14, 2017

Dockets Management Branch (HFA-305)

Food and Drug Administration

5630 Fishers Lane, Rm. 1061

Rockville, MD 20852

Re: Docket No. FDA-2016-D-4460: Multiple Endpoints in Clinical Trials

Dear Sir/Madam:

The Biotechnology Innovation Organization (BIO) thanks the Food and Drug Administration

(FDA) for the opportunity to submit comments on the Draft Guidance “Multiple Endpoints in

Clinical Trials” (Draft Guidance).

BIO is the world's largest trade association representing biotechnology companies, academic

institutions, state biotechnology centers and related organizations across the United States

and in more than 30 other nations. BIO members are involved in the research and

development of innovative healthcare, agricultural, industrial, and environmental

biotechnology products.

This important Draft Guidance was well written and provides instrumental guidance and

recommendations to handle major multiplicity problems in clinical trials. BIO believes that

the Draft Guidance is a very helpful document to Sponsors. However, we note that the Draft

Guidance fails to address a few points that are worthy of attention:

multiplicity adjustments at the interim analysis (either directly in this Draft Guidance or

via reference to ICH E9 or the Guidance for Industry Adaptive Design Clinical Trials for

Drugs and Biologics);

multiplicity adjustment on safety endpoints when assessing important safety signals as

part of the objectives of the trial (e.g., with p-values reported) to control the overall

false discovery rate of safety signals;

elucidation of the calculation methods to be used with the truncated Holm and truncated

Hochberg procedures (e.g., for the truncated Hochberg example, the α passed to the

next level); and

basic principles, such as closed test or partition principles, which many of the multiple

comparison methods in the guidance are based upon.

BIO further believes that it would be helpful if the Draft Guidance stated at the beginning,

perhaps more clearly and emphatically than it does now, the areas that are mentioned in

other sections as being out of scope. It would also be helpful to explain why these areas are

outside of the scope. For example, the Draft Guidance states that multiplicity in interim

analysis is not within the scope of the document (lines 180-184). This is something that

should also be stated in the introduction. Simulation based multiplicity adjustment is

another area that is not included in the guidance. If related topics are outside the scope but

they have been the subject of previous FDA guidance, such guidance should be referenced.

BIO Comments on Multiple Endpoints in Clinical Trials FDA Docket: FDA-2016-D-4460, March 14, 2017, Page 2 of 21

We note that in multiple places in the document, reference is made to the ‘endpoint’ when

we believe that reference is intended to be made to the hypothesis or the hypothesis test

(e.g., lines 252-253, 491-492, and 885). This language is likely sufficiently clear for many

readers of the guidance. However, FDA may consider correcting this language throughout

when revising the guidance. Alternatively, a statement early in the document can be

inserted which explains this use of terminology as interchangeable in the document. This

would help improve the clarity of the Draft Guidance.

Further, the Draft Guidance does not clearly indicate whether the Hochberg procedure is

recommended in the case of more than two correlated endpoints. Under the multivariate

central limit theorem, the joint density of the multiple test statistics for three non-negatively

correlated endpoints will follow a multivariate normal distribution in confirmatory trials

where large sample sizes are typically planned. Therefore, the Hochberg procedure

applicable to two correlated endpoints should apply to more than two non-negatively

correlated endpoints. It would be helpful if the Draft Guidance discusses and clarifies this

topic.

Additionally, we believe that “Section IV Statistical Methods” can be improved upon via

incorporation of alpha recycling (see below citations1). We suggest that FDA consider adding

a statement that incorporation of alpha recycling, as shown in the appendix on graphical

approaches, may provide extensions of the methods described in this section. We suggest

that FDA consider adding a paragraph referring to the appendix for graphical approaches

that can be used to improve the methods described in this section.

Regarding the graphical approach, BIO notes that many testing procedures described in the

Draft Guidance, including the Bonferroni method, the Holm procedure, the fixed-sequence

method, and the Fallback method, are special cases of multiple testing procedure. We

suggest FDA make the connection between the graphical approach and these special cases.

The Draft Guidance also considers the graphical approach as a visual presentation tool

instead of recognizing it as an extension of many commonly used statistical methods. It is

an important multiple testing procedure. Given the importance of the graphical approach

and its connection to other methods, including gate keeping testing strategies, it is worth

having a small section in the main text body (similar to re-sampling test method), yet still

retain the details in the appendix. Discussing it solely in the appendix will convey to some

readers that it is a less favored method. It provides uniformly more power across many

statistical situations. It also provides much greater flexibility in organizing the hypothesis

testing and the amount of alpha available to each hypothesis test. It also allows the clinical

importance of each endpoint to be taken into account. Finally, the graphical approach is also

extremely valuable as a communication tool. It communicates complex, hypothesis testing

plans that cannot be clearly communicated in text. However, because of its extensive

flexibility, it is not always clear which endpoints are the primary endpoints and which ones

are the secondary endpoints. The guidance should encourage sponsors that when they are

1 Burman CF, Sonesson C, Guilbaud O. A recycling framework for the construction of Bonferroni-based multiple tests. Statistics in Medicine. 2009; 28:739--761. Bretz F, Maurer W, Brannath W, Posch M. A graphical approach to sequentially rejective multiple test procedures. Statistics in Medicine} 2009; 28:586--604. Millen B, Dmitrienko A. Chain procedures: A class of flexible closed testing procedures with clinical trial applications. Statistics in Biopharmaceutical Research 2011; 3:14--30.

BIO Comments on Multiple Endpoints in Clinical Trials FDA Docket: FDA-2016-D-4460, March 14, 2017, Page 3 of 21

using the graphical approach, that they should clearly state the intended role of endpoints

(i.e., primary basis for approval vs. supportive). This will help to prevent misunderstandings

between the sponsor and the Agency about what is necessary for approval.

While we understand that the guidance cannot cover every statistical method that could be

relevant for multiple endpoints, we do believe it would be helpful if the guidance provided

some information about the criteria used to decide which methods to include. For instance,

a variety of innovative study designs are receiving greater attention, such as Umbrella

designs, Basket designs, and Platform designs. They are considered to have great potential

for making drug development more efficient. FDA should consider mentioning these in the

guidance. Additionally, it would be helpful if FDA provided some information about what

distinguishes the methods that were included in the main document versus the appendix.

Finally, we believe that an extension of this guidance or an additional guidance which more

broadly discusses the problem of “multiplicity” would be beneficial. In particular, we believe

that additional guidance regarding the interpretation of subgroup analyses would be helpful

in this guideline both for trials that did or did not meet their primary objective in the overall

population, respectively. Other topics of interest that could be addressed in an additional

guideline include analyses of multiple time-points (e.g., group-sequential trials), patient

selection (biomarker) subgroups, and multi-arm trials including trials with multiple doses

which may be modelled.

BIO appreciates this opportunity to submit comments on the Draft Guidance “Multiple

Endpoints in Clinical Trials”. We provide additional specific, detailed comments to improve

the clarity of the Draft Guidance in the following chart. We would be pleased to provide

further input or clarification of our comments, as needed.

Sincerely,

/S/

Cartier Esham, Ph.D.

Executive Vice President, Emerging Companies Section &

Vice President, Science & Regulatory Affairs

Biotechnology Innovation Organization

BIO Comments on Multiple Endpoints in Clinical Trials FDA Docket: FDA-2016-D-4460, March 14, 2017, Page 4 of 21

SPECIFIC COMMENTS

SECTION ISSUE PROPOSED CHANGE

I. INTRODUCTION

II. BACKGROUND AND SCOPE

A. Introduction to Study Endpoints

B. Demonstrating the Study Objective of Effectiveness

Line 130: The null hypotheses is presented as 2-sided, but the

alternative hypothesis is presented as 1-sided.

BIO asks FDA to make the null hypothesis 1-sided to be

consistent with the alternative hypothesis in this paragraph

and the other one refer to Line 142 to Line 145 in the last

paragraph of the section.

C. Type I Error

Lines 154-156,

158-162:

A definition of Type I error probability for the 2-sided

hypothesis tests is given at Lines 154-156 without

clearly stating this is a definition applied to the 2-

sided hypothesis tests. The same definition is

repeated at Lines 158-160 followed by a slightly

different definition of Type I error rate for 1-sided

hypothesis tests.

BIO suggest that FDA:

(1) re-write the sentence at Lines 154-156 to read as

“Under two-sided hypothesis tests, the probability of

concluding that there is a difference (beneficial or

harmful) between the drug and control when there is

no difference, is called the Type I error probability or

rate, denoted as alpha (α). and

(2) delete the first sentence in the 2nd paragraph of

Section II.C (Lines 158-160) as it repeats the same

definition that has been given at Lines 154-156.

Lines 168-170: The rationale as to why the use of 2-sided test would

provide strong assurance against the possibility of a

false-positive result (i.e., no more than 1 chance in

40), is a bit unclear (especially for non-statisticians).

BIO suggests re-writing this sentence to read:

“Use of a two-sided test with an alpha of 0.05 generally

allocates the alpha in a symmetric way, such also ensures

that the probability of falsely concluding the drug effect in

each of the two possible directions (benefit or harm) is no

more than 2.5 percent. benefit This ensures that the

BIO Comments on Multiple Endpoints in Clinical Trials FDA Docket: FDA-2016-D-4460, March 14, 2017, Page 5 of 21

SECTION ISSUE PROPOSED CHANGE

probability of falsely concluding drug benefit when there is

none is no more than approximately 2.5 percent (1 chance

in 40).”

Lines 170-172: The Draft Guidance reads, “…especially when

substantiated by another study or other confirmatory

evidence.”

BIO notes that this is not always the case, and it is

preferable to acknowledge that single studies may be

acceptable.

BIO suggests editing the text to read:

“....especially when substantiated by another study or other

confirmatory evidence. It should be noted that for orphan

diseases or diseases with high unmet medical needs, a single

study may be acceptable for regulatory approval.”

D. Relationship Between the Observed and True Treatment Effects

Lines 203-204: The Draft Guidance reads:

“An essential element of type I error rate control is

the prospective specification of:

all endpoints that will be tested and”

Since hypothesis testing is sometimes a part of

evaluating exploratory objectives which are out of

scope from a multiple testing perspective, we

recommend clarifying that the statement applies to

tests of ‘primary and secondary hypothesis tests’

rather than ‘all’ tests.

In order to clarify the Draft Guidance, BIO suggests editing

the text to read:

“An essential element of type I error rate control is the

prospective specification of:

all endpoints primary and secondary hypotheses that

will be tested and”

Lines 207-209: The Draft Guidance discusses the analysis plan for

multiple endpoints studies.

These lines in the draft guidance attempt to clarify

details needed in an analysis plan to pre-specify and

communicate a multiple testing plan. To more

BIO suggests replacing the text in this section with the

following:

The analysis plan should describe the multiple testing

procedure for the hypotheses being tested. This may include

specifying the ordering of testing the hypotheses, the initial

BIO Comments on Multiple Endpoints in Clinical Trials FDA Docket: FDA-2016-D-4460, March 14, 2017, Page 6 of 21

SECTION ISSUE PROPOSED CHANGE

completely accommodate the broad range of methods

to be conveyed, we suggest additions to this

statement which will be helpful for both reviewers

and practitioners in industry.

alpha allocation to the hypotheses, and the alpha

propagation rules (i.e., how alpha is passed from one

hypothesis test to another). These details may be

communicated in text and/or pictorials/graphics. (See

appendix for description of a graphical approach of

representing multiple testing procedures. Other conventions

are available as well. See, for example, Khordzhakia et al

(2012) and Dmitrienko et al (2013).) An alternative means

of describing the multiple testing procedure is to provide the

closed testing representation or partition testing

representation of the procedure. Any of these approaches is

sufficient to fully specify and communicate the details of the

procedure.

Lines 225-230: The Draft Guidance states, “The narrower the

confidence interval, and the further away its lower

bound is from the null hypothesis of no treatment

effect (T-C = 0 or T/C = 1), the more confident we

are of both the magnitude and existence of the

treatment effect. Generally, the farther the lower

bound of the confidence interval is from zero (or 1),

the more persuasive (smaller) the p-value is and the

lower the likelihood that the effectiveness finding was

a chance occurrence.”

Only the lower bound of CI’s is mentioned but in many

situations, the upper bound is of interest (for example, for

hazard ratios of survival endpoints). BIO suggests editing

the text to read:

“The narrower the confidence interval, and the further away

its lower or upper bound, respectively, (depending on the

direction of the effect of interest) is from the null hypothesis

of no treatment effect (T-C = 0 or T/C = 1), the more

confident we are of both the magnitude and existence of the

treatment effect. Generally, the farther the lower or upper

bound of the confidence interval is from zero (or 1), the

more persuasive (smaller) the p-value is and the lower the

likelihood that the effectiveness finding was a chance

occurrence.”

Lines 243-246: The Draft Guidance discusses confidence intervals for

testing and reads,

The text referencing confidence intervals for testing here

may be confusing to readers as referring to multiplicity-

adjusted confidence sets, which is generally out of scope for

BIO Comments on Multiple Endpoints in Clinical Trials FDA Docket: FDA-2016-D-4460, March 14, 2017, Page 7 of 21

SECTION ISSUE PROPOSED CHANGE

“…it is critical to ensure that confidence intervals

appropriately reflect multiplicity of hypothesis tests.”

the document. Use of a confidence interval for testing does

not present a special case to be addressed separately. The

guidance on testing is wholly adequate. As such, BIO

suggests removing these lines to avoid confusion or have the

following revision:

“…it is critical to ensure that the confidence level of

confidence intervals appropriately reflect multiplicity of

hypothesis tests.”

E. Multiplicity

Lines 269-271: The stated type I error rate of 0.1 is approximate.

The exact error rate is given just prior to this

statement. The proposed text corrects this minor

error.

BIO suggests editing the text to read:

“Without correction, the chance of making a type I error for

the study as a whole would be approximately 0.1 and…”

Lines 309-310: The text states that “post hoc analyses by

themselves cannot establish effectiveness.” However,

there are cases where the agency has exercised

flexibility in this regard. For example, Pravagard PAC

was approved on the basis of a post hoc meta-

analysis of individual patient data from previous

clinical trials of the same compound. Hence, this

statement may have to be modified to accommodate

unusual situations.

BIO suggests editing the text to read:

“Consequently, post hoc analyses by themselves generally

cannot establish effectiveness.”

III. MULTIPLE ENDPOINTS: GENERAL PRINCIPLES

A. The Hierarchy of Family of Endpoints

Lines 391-393: The Draft Guidance states, “The collection of all

secondary endpoints is called the secondary endpoint

family. Secondary endpoints are those that may

provide supportive information about a drug’s effect

BIO also asks FDA to please provide additional detail and

examples here or in Lines 327-334 on the requirements for

endpoints, beyond the primary endpoint, that can be

included in physician labeling.

BIO Comments on Multiple Endpoints in Clinical Trials FDA Docket: FDA-2016-D-4460, March 14, 2017, Page 8 of 21

SECTION ISSUE PROPOSED CHANGE

on the primary endpoint or demonstrate additional

effects on the disease or condition.”

Endpoints “that may provide supportive information

about a drug’s effect on the primary endpoint” and

endpoints that “demonstrate additional effects on the

disease or condition” seem quite different, and it is

not clear why the former should be considered a

secondary endpoint (i.e., require multiplicity control).

In addition, this seems inconsistent with lines 327-

337 which imply that such endpoints (i.e., those that

describe other attributes of a drug’s effects) can be

included in physician labeling without multiplicity

control. Therefore, the distinction between endpoints

that do and do not require multiplicity control is

unclear.

Lines 409-413: The Draft Guidance states, “Moreover, the Type I

error rate should be controlled for any preplanned

analysis of pooled results across studies; pooled

analyses are rarely conducted for the planned

primary endpoint, but are sometimes used to assess

lower frequency events, such as cardiovascular

deaths, where the individual trials used a composite

endpoint, such as death plus hospitalization.

Statistical testing strategies to accomplish this are

discussed in section IV.”

While the Draft Guidance indicates that discussions

on preplanned pooled analysis to assess lower

frequency events as secondary endpoints and

methods to control Type I error, whether separately

These lines refer to type I error rate control for analyses

based on pooled results across studies. BIO asks FDA to

provide more detail, if possible, on what is expected in this

setting.

BIO Comments on Multiple Endpoints in Clinical Trials FDA Docket: FDA-2016-D-4460, March 14, 2017, Page 9 of 21

SECTION ISSUE PROPOSED CHANGE

or using a pre-planned alpha allocation, are discussed

in section IV, this does not appear to be the case.

Lines 431-433: The draft Guidance reads, “…becomes increasingly

small as the number of endpoints increases”

This statement is not accurate because the statement

is not necessary true for some multiplicity adjustment

methods. For example, in the fixed-sequence

procedure, the full alpha is passed to the next

endpoint if the previous endpoint is statistically

significant at the 0.05 level. Therefore, the chance of

demonstrating an effect on a series of sequential

endpoints depends on the method used for

multiplicity adjustment.

BIO suggests the text be edited to read:

“could becomes increasingly small as the number of

endpoints increases.”

Lines 443-445: The Draft Guidance reads, “The study’s likelihood of

avoiding Type II error (1-β), if the drug actually has

the specified effect, is called study power. The

desired power is an important factor in determining

the sample size”

The type II error is β, not 1-β.

The study power is 1-β.

Power for the secondary endpoints is not typically

considered as power loss after passing the primary

endpoints. Power should be associated with the

sample size for the primary endpoint.

BIO suggest the editing the text to read:

“The study’s likelihood of avoiding Type II error (1-β), if the

drug actually has the specified effect, is called study power

(1-β). The desired power is an important factor in

determining the sample size, especially for the primary

endpoints.”

B. Type II Error Rate and Multiple Endpoints

BIO Comments on Multiple Endpoints in Clinical Trials FDA Docket: FDA-2016-D-4460, March 14, 2017, Page 10 of 21

SECTION ISSUE PROPOSED CHANGE

Lines 483-484: The draft Guidance states, “The loss of power may

not be so severe when the endpoints are correlated

(i.e., not fully independent)”.

This statement is not accurate because the loss of

power may be severe for weak correlated endpoints

or negatively correlated endpoints if one endpoint is

successful. The statement is true for endpoints with

strong positive correlation. Therefore, we revised the

statement to make it accurate.

In addition, the phrase “(i.e., not fully independent)”

is problematic for this statement. If the correlation of

endpoints is not high, then the power loss may be

severe for the endpoint with small treatment effect

size. Thus, we suggest deleting the parenthetical

statement.

BIO suggests editing the text to read:

“The loss of power may not be so severe when the endpoints

are strongly positively correlated (i.e., not fully

independent).”

C. Types of Multiple Endpoints

Line 507 BIO notes that the term “co-primary endpoints” is

used specifically for the case of all-of-N, must be

statistically significant. In some therapeutic areas

that term is also used for 1 of N.

BIO thinks it would be useful for FDA to mention the use of

the term “co-primary endpoints” for 1 of N in this discussion.

BIO also suggests the FDA clarifies that in this Guidance, the

term “co-primary endpoints” is exclusively used for the case

of all-of-N.

Lines 554-558: It is possible to test for co-primary endpoints in a

manner which controls the overall type I error at the

required alpha level (e.g., 0.05) yet does not require

each endpoint to be tested at the 0.05 level. See

Kordzhakia et al, 2010, for example.

BIO asks FDA to consider broadening the language on co-

primary endpoints to allow for the possibility mentioned:

When using co-primary endpoints, however there is

generally only one result that is considered a study success,

namely, that all of the separate endpoints are statistically

significant. With this approach, there is no opportunity for

BIO Comments on Multiple Endpoints in Clinical Trials FDA Docket: FDA-2016-D-4460, March 14, 2017, Page 11 of 21

SECTION ISSUE PROPOSED CHANGE

It should be permissible to select significance levels

for testing the individual endpoints in any way that

controls the overall Type I error at the required level

α, even if one or more of these individual significance

levels are greater than α. The set of admissible

individual significance levels will depend on the

number of co-primary endpoints as well as the

specific formulation of the null hypothesis, and the

final choice should be justified by an objective

demonstration of overall Type I error control.

inflation of the type I error rate; rather the impact of co-

primary endpoint testing is to increase the type II error rate.

While statistical significance for each co-primary endpoint is

sufficient to satisfy the requirement of overall type I error

control, it is also possible to maintain overall type I error

control using other significance levels for the individual

hypotheses. (See Kordzhakia et al, 2010, for example.)

Such an approach may be considered in some cases where

power is greatly reduced when simply testing each co-

primary endpoint at level alpha.

Lines 564-566: The Draft Guidance states, “Relaxation of alpha is

generally not acceptable because doing so would

undermine the assurance of an effect on each disease

aspect considered essential to showing that the drug

is effective in support of approval.”

When co-primary endpoints are required yet not

highly correlated (e.g., histological evidence and

symptom improvement), not relaxing alpha for each

individual co-primary component may result in much

reduced study-wise Type I error rate and

unnecessary large sample size, increased drug

development timeline and cost.

BIO suggests that FDA consider clarifying the ability of

sponsors to engage in application specific discussions.

Lines 596-631: This section on composite endpoints may touch on

recent developments, especially analysis that takes

clinical importance into account, such as win-ratio

analysis or weighted analysis.

BIO suggests FDA consider elaborating on some of these

new developments and analyses.

Lines 608-614: “An important reason for using a composite endpoint

is that the incidence rate of each of the events may

BIO suggests editing the text to read:

BIO Comments on Multiple Endpoints in Clinical Trials FDA Docket: FDA-2016-D-4460, March 14, 2017, Page 12 of 21

SECTION ISSUE PROPOSED CHANGE

be too low to allow a study of reasonable size to have

adequate power; the composite endpoint can provide

a substantially higher overall event rate that allows a

study with a reasonable sample size and study

duration to have adequate power. Composite

endpoints are often used when the goal of treatment

is to prevent or delay morbid, clinically important but

uncommon events (e.g., use of an anti-platelet drug

in patients with coronary artery disease to prevent

myocardial infarction, stroke, and death).”

BIO notes that another reason for composite

endpoints is when a more serious (but less common)

event occurs in the absence of a documented more

common (but less serious) event, for example deaths

in the absence of disease progression or relapse. In

such cases, the more severe event should not be

ignored.

“An important One possible reason for using a composite

endpoint is that the incidence rate of each of the events may

be too low to allow a study of reasonable size to have

adequate power; the composite endpoint can provide a

substantially higher overall event rate that allows a study

with a reasonable sample size and study duration to have

adequate power. Composite endpoints are often used when

the goal of treatment is to prevent or delay morbid, clinically

important but uncommon events (e.g., use of an anti-

platelet drug in patients with coronary artery disease to

prevent myocardial infarction, stroke, and death).”

Lines 678-679: The Draft Guidance states, “Study power can be

adversely affected, however, if there is limited

correlation among the endpoints.”

It is not clear to BIO whether the document is

referring to the issue of reverse multiplicity or

differences in underlying effect sizes.

BIO suggests the FDA provides clarification on this point.

Lines 689-691: BIO notes that it is frequently meaningful to add

“worse outcomes” (i.e., rare but clinically critical

endpoints) as components to the primary endpoints

to avoid interpretational issues due to competing

risks. We suggest comment or acknowledgement of

this principle.

BIO suggests editing the text to read:

“For many serious diseases, there is an endpoint of such

great clinical importance that it is unreasonable not to collect

and analyze the endpoint data; the usual example is

mortality or major morbidity events (e.g., stroke, fracture,

BIO Comments on Multiple Endpoints in Clinical Trials FDA Docket: FDA-2016-D-4460, March 14, 2017, Page 13 of 21

SECTION ISSUE PROPOSED CHANGE

pulmonary exacerbation). Even if relatively few of these

events are expected to occur in the trial, they may be

included it is frequently beneficial from an interpretational

perspective to include them in a composite endpoint (see

section III.C.3) and they may also be designated as a

planned secondary endpoint to potentially support a

conclusion regarding effect on that separate clinical

endpoint, if the effect of the drug on the composite primary

endpoint is demonstrated.”

D. The Individual Components of Composite Endpoints

Lines 730-733: The Draft Guidance states, “One approach considers

only the initial event in each patient. This method

displays the incidence of each type of component

event only when it was the first event for a patient.

The sum of the first events across all categories will

equal the total events for the composite endpoint.”

BIO asks the Agency to consider clarifying that sponsors

have additional options regarding analysis based on only the

initial event in each patient, due to a serious methodological

issue. For example, consider the analysis of ESRD in RENAAL

as described in the document. The numbers for ESRD under

“Decomposition of the primary endpoint” represents the

numbers of patients who had ESRD that was not preceded

by Doubling of Serum Creatinine. A reduction in this number

is necessarily accompanied by an equal increase in the sum

of these two categories of patients: 1) Those without ESRD

at all (a favorable outcome); and 2) Those with Doubling of

Serum Creatinine followed by ESRD (an unfavorable

outcome). In other words, the approach discussed by the

FDA essentially creates a composite endpoint that comprises

both favorable and unfavorable outcomes. This violates what

should be an absolute criterion for creation of composite

endpoints, because it is unclear whether an increase or a

decrease in this endpoint would be of benefit to the patient.

Lines 741-767: The example is very long without conclusions and

may not provide any helpful information.

BIO Comments on Multiple Endpoints in Clinical Trials FDA Docket: FDA-2016-D-4460, March 14, 2017, Page 14 of 21

SECTION ISSUE PROPOSED CHANGE

Line 757 (Table

1):

The footnote says “Hazard ratio from Cox

proportional hazards time-to-event analysis”.

However, as competing risks are present for these

analyses, it should be clarified that this refers to

models for the cause-specific hazards functions.

Moreover, in the presence of competing risks,

alternative regression models such as the Fine and

Gray would also be applicable.

BIO believes that this issue merits further discussion

and possibly a reference to a paper on competing

risks and multi-stage models. E.g. The “Tutorial in

biostatistics: competing risks and multi-state

models.” published in Statistics in Medicine by

Geskus et al (2007).

More generally, we think that further guidance on the

topic of competing risks, which is only briefly

mentioned in Lines 770-776, would be beneficial.

BIO suggests editing the text to read:

“Hazard ratio from cause-specific Cox proportional hazards

time-to-event analysis accounting for competing events. Of

note, component endpoints could alternatively be analyzed

on the cumulative incidence scale using the Fine and Gray

regression model.”

Lines 779-784: The Draft Guidance refers to “decomposition

analyses” but not define the term.

BIO finds the meaning of “decomposition” in this context is

not clear. BIO asks FDA to please clarify.

IV. STATISTICAL METHODS

A. Type I Error Rate for a Family of Endpoints and Conclusions on Individual Endpoints

B. When the Type I Error Rate is Not Inflated or When the Multiplicity Problem is Addressed Without Statistical Adjustment or by Other

Methods

Lines 933-937: The Draft Guidance reads, “but it is difficult to

estimate the increase in error rate because the

results of these different analyses are likely to be

similar and it is unclear how many choices could have

BIO believes that the guidance should clearly state that one

analysis method must be the primary method and pre-

specified so that a biased choice cannot be made. The

BIO Comments on Multiple Endpoints in Clinical Trials FDA Docket: FDA-2016-D-4460, March 14, 2017, Page 15 of 21

SECTION ISSUE PROPOSED CHANGE

been made. As with other multiplicity problems,

prospective specification of the analysis method will

generally eliminate the concern about a biased

(result-driven) choice.”

difficulty in estimating the increase in Type 1 error rate is

irrelevant. As such, BIO suggests editing the text to read:

“but it is difficult to estimate the increase in error rate

because the results of these different analyses are likely to

be similar and it is unclear how many choices could have

been made. As with other multiplicity problems, prospective

specification of the analysis method will generally eliminate

the concern about a biased (result-driven) choice. One

method should be pre-specified as the primary analysis

method to eliminate the possibility of a result based on a

biased choice.”

C. Common Statistical Methods for Addressing Multiple Endpoint-Related Multiplicity Problems

Line 978: It will be helpful for both reviewers and practitioners

in industry to provide reference to general

construction of multiple testing procedures in addition

to the ones presented in the document.

BIO suggests adding the following text to this part of the

Statistical Methods section:

It is, of course, impossible to include discussion or examples

of all potential multiple testing procedures applicable to

clinical trials. However, two important principles which

allows for the construction of multiple testing procedures

which provide strong type I error are: the closed testing

principle (or closure principle) and the partitioning principle.

These principles may be used to develop procedures beyond

those outlined below.

Line 982-983: The Draft Guidance states, “Single-step procedures

tend to cause loss of study power, so that sample

sizes need to be increased in comparison to sample

sizes needed for a single-endpoint study.”

BIO asks FDA to clarify the intended meaning of this section.

BIO Comments on Multiple Endpoints in Clinical Trials FDA Docket: FDA-2016-D-4460, March 14, 2017, Page 16 of 21

SECTION ISSUE PROPOSED CHANGE

It is not clear that this is necessarily the case. For

example, if there are two independent endpoints, A

and B, each with 90% power at the 5% significance

level, the power for a single-step Bonferroni

procedure (i.e., the probability that either one of the

two would achieve a p-value < 0.025) would be

approximately 97%, which is greater than the power

for either as a single endpoint. The FDA might have

intended to say that a single-step test of hypotheses

A and B has less power for showing an effect on A

than a test of A as a single endpoint.

Lines 1008-1009: The Draft Guidance states, “The most common form

of the Bonferroni method divides the available total

alpha (typically 0.05) equally among the chosen

endpoints.”

BIO asks FDA to clarify that the typical alpha=0.05 is two-

sided:

“The most common form of the Bonferroni method divides

the available total alpha (typically 0.05 two-sided) equally

among the chosen endpoints.”

Lines 1031-1033: The Draft Guidance reads, “When a multiple-arm

study design is used (e.g., with several dose-level

groups), there are methods that take into account

the correlation arising from comparing each

treatment group to a common control group.”

BIO notes that the language in this section is the Dunnett

method. However, FDA does not seem to refer to is as such.

The method’s name is mentioned later (line 1394), so it

should also be mentioned here. Making clear to the reader

that this is the Dunnett method will provide more clarity to

reader. A such, BIO suggests editing the text to read:

“When a multiple-arm study design is used (e.g., with

several dose-level groups), there are methods that take into

account the correlation arising from comparing each

treatment group to a common control group (i.e., Dunnett

method).”

BIO Comments on Multiple Endpoints in Clinical Trials FDA Docket: FDA-2016-D-4460, March 14, 2017, Page 17 of 21

SECTION ISSUE PROPOSED CHANGE

Lines 1053-1111

(Section IV C2

The Holm

Procedure)

BIO finds it is confusing when use of “endpoints 1, 2,

3, 4”, don’t correspond to the order in which they are

tested. Specifically, on line 1093, “endpoint 3” is the

second endpoint tested.

BIO suggests naming the endpoints as A, B, C, and D for

clarity.

Lines 1078-1080: The Draft Guidance reads, “The procedure stops,

however, whenever a step yields a non-significant

result. Once an ordered p-value is not significant, the

remaining larger p-values are not evaluated and it

cannot be concluded that a treatment effect is shown

for those remaining endpoints.”

BIO notes that the adjusted p-values for all

comparisons can be calculated, it is just all

comparisons after the first non-significant result are

non-significant. This also applies to the text in lines

1225-1226.

BIO asks FDA to please clarify whether testing stops or

whether further tests are not considered statistically

significant.

Lines 1238-1239: The Draft Guidance states, “The Holm test would not

find significant effects for additional endpoints either,

unless the p-value for endpoint A was p < 0.025.”

BIO notes that both "unless pA < 0.025" is equally as

true as "unless pC < 0.025"? While we believe that

neither statement is necessary, just giving one

causes confusion.

To reduce confusion BIO recommends either changing the

text to read:

“The Holm test would not find significant effects for

additional endpoints either, unless the p-value for either

endpoint A or C was p < 0.025.”

Or removing the latter section of the sentence altogether:

“The Holm test would not find significant effects for

additional endpoints either, unless the p-value for endpoint A

was p < 0.025.”

Lines 1240-1241: The Draft Guidance discusses failed studies.

However, BIO notes that a lack of statistical

significance does not make a study a failed study.

To ensure accuracy in the Draft Guidance, BIO recommends

editing the text to read:

BIO Comments on Multiple Endpoints in Clinical Trials FDA Docket: FDA-2016-D-4460, March 14, 2017, Page 18 of 21

SECTION ISSUE PROPOSED CHANGE

“…it would be an entirely failed study have failed to detect

significant effects…”

Line 1315: BIO believes that the Fallback procedure can be

uniformly improved (which means it is uniformly

more powerful than its simple variation). It is

important to point this out when describing the

method in guidance document.

BIO suggests adding the following sentence to this section:

"The fallback procedure can be uniformly improved in terms

of power. Refer to Appendix Figure A4 for examples."

Line 1390: It is important to point out to readers that, even with

different names and appearances, some common

procedures lead to the same results.

BIO suggests adding the following sentence to this section:

"It is worth noting that, in an important common setting

(multiple endpoints, multiple doses), even with different

appearances, the parallel gatekeeping procedure, the

graphical approach, and the partitioning approach lead to

essentially the same testing results."

Lines 1432-1435: The Draft Guidance states, “The endpoint specific

alpha levels for the truncated Holm are then

constructed by combining the endpoint specific alpha

levels of the two methods with a “truncation fraction”

of f, whose value between zero and one is selected in

advance.”

BIO asks FDA to consider additional guidance for choosing f,

the truncation fraction, e.g. by maximizing the probability of

rejecting at least one true null hypothesis in each family, as

suggested by Dmitrienko, Tamhane and Wiens, 2008.

Lines 1449-1453: The Draft Guidance states, “i. Unused alpha = 0.05, if

all three tests are successful;

ii. Unused alpha = (0.05 –a3) = 0.05 - 0.0333 =

0.0167, if the first two tests are successful, but the

last one is not;

iii. Unused alpha = (0.05 – 2a2) = 0.05 -2(0.0208) =

0.0084, if the first test is successful, but the other

two tests are not.”

BIO finds the this portion of the Draft Guidance to be a bit

confusing to understand the calculation of the significance

level for family 2. For clarity we suggest using a more

generic formula such as:

unused alpha=(1-f)*0.05*r1/k1, where r1 is the number of

null hypotheses rejected in primary family and k1 is the total

number of null hypotheses in primary family.

BIO Comments on Multiple Endpoints in Clinical Trials FDA Docket: FDA-2016-D-4460, March 14, 2017, Page 19 of 21

SECTION ISSUE PROPOSED CHANGE

Lines 1553-1556: The Draft Guidance states, “Therefore, once the

result for H1 is significant at level a (i.e., the

treatment is non-inferior to control for endpoint one

at level a), testing proceeds to the hypotheses H2 and

H3 in group two with the alpha that was not used

within family one, which in this case would be the

overall study alpha.”

BIO finds the wording in this section misleading. The

significance of the results for NI trials is not usually

discussed, as the confidence interval approach is used. As

such we suggest editing the text to read:

“Therefore, once the result for H1 is rejected at level alpha is

significant at level a (ie, the treatment is non-inferior to

control for endpoint one at level a), testing proceeds to the

hypotheses H2 and H3 in group two with the alpha that was

not used within family one, which in this case would be the

overall study alpha.”

Lines 1573-1574: This is the only place where the graphic approach is

mentioned in the guideline besides Appendix A.

BIO suggests adding a section to show the graphic approach

as a statistical method for multiplicity control because of its

important features such as alpha passing back and alpha

propagation.

Line 1576: BIO finds this method difficult to understand.

BIO suggests providing an example for re-sampling based

multiplicity-testing procedure to explain how multiplicity

control is done.

Lines 1587-1595: BIO finds this paragraph to be confusing. Normal

approximation requires large sample size, not re-

sampling which requires minimum assumptions to

justify the methods. By the same logic, Type I error

rate may occur for normal approximation, but not as

much for re-sampling methods. It is “modeling” that

requires strong assumptions.

Please see comment above in line 1578-1585.

Lines 1598-1599: BIO believes that discouraging any re-sampling based

approaches as primary analysis methods is rather

restrictive and does not give any room for

BIO suggests the following alternative wording:

BIO Comments on Multiple Endpoints in Clinical Trials FDA Docket: FDA-2016-D-4460, March 14, 2017, Page 20 of 21

SECTION ISSUE PROPOSED CHANGE

methodological developments that may occur over

the lifetime of this guidance document.

“Because of this, resampling methods are not recommended

used as primary analysis methods for adequate and well-

controlled trials in drug development should robustly have

demonstrated Type I error rate control.”

Line 1602: BIO believes it would be helpful to add a short

discussion regarding re-samples on residuals.

BIO suggests adding the following text at the end of this

section:

“Note, however, if re-sampling is not done on the residuals,

instead permuting treatment labels or outcome measures for

example, then strong distributional assumptions are needed

for FWER control.”

Line 1602: BIO notes that the Permutation test may not control

FWER. Since permutation test is included in this

guidance (line 1587), it is important to warn readers

about the possibility of failing to control FWER when

using the permutation test.

BIO suggests adding the following sentence:

“It is important to note that for bootstrap or permutation,

strong distributional assumptions may be required for FWER

control. For example, permuting treatment groups,

biomarkers (in subgroup setting), or the outcome measure

(instead of residuals after modeling) does not necessarily

control FWER.”

V. CONCLUSION

Lines 1618-1620: The Draft Guidance states, “this guidance is intended

to clarify when and how multiplicity due to multiple

endpoints should be managed to avoid reaching such

false conclusions.”

Overall the guidance addresses multiple testing of

hypotheses due to multiple testing of hypotheses,

whether at different timepoints, for different endpoint

To ensure accuracy in the guidance, BIO suggests editing the

concluding text to read:

“this guidance is intended to clarify when and how

multiplicity due to multiple endpoints hypothesis tests should

be managed to avoid reaching such false conclusions.”

BIO Comments on Multiple Endpoints in Clinical Trials FDA Docket: FDA-2016-D-4460, March 14, 2017, Page 21 of 21

SECTION ISSUE PROPOSED CHANGE

measures, at different looks (interim or final), for

different populations, etc. BIO believes that the

concluding language regarding multiple endpoints

does not adequately reflect this.

APPENDIX: THE GRAPHICAL APPROACH

Line 1779: Correct typo of “alphas” to “alpha”.

Line 1882: The text reads “ash shown in Figure A5(a),” BIO suggests editing the text to read:

“as shown in Figures A5(a) and A5(b),”

Line 1922: BIO notes that the alpha for H3 in Figure A5 (c) should be

epsilon*alpha2.